Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38585793

RESUMO

Transposable elements (TEs) make up the bulk of eukaryotic genomes and examples abound of TE-derived sequences repurposed for organismal function. The process by which TEs become coopted remains obscure because most cases involve ancient, transpositionally inactive elements. Reports of active TEs serving beneficial functions are scarce and often contentious due to difficulties in manipulating repetitive sequences. Here we show that recently active TEs in zebrafish encode products critical for embryonic development. Knockdown and rescue experiments demonstrate that the endogenous retrovirus family BHIKHARI-1 (Bik-1) encodes a Gag protein essential for mesoderm development. Mechanistically, Bik-1 Gag associates with the cell membrane and its ectopic expression in chicken embryos alters cell migration. Similarly, depletion of BHIKHARI-2 Gag, a relative of Bik-1, causes defects in neural crest development in zebrafish. We propose an "addiction" model to explain how active TEs can be integrated into conserved developmental processes.

2.
Front Cell Dev Biol ; 12: 1324584, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38655067

RESUMO

ASCL1 is a transcription factor that directs neural progenitors towards lineage differentiation. Although many of the molecular mechanisms underlying its action have been described, several of its targets remain unidentified. We identified in the chick genome a putative enhancer (cE1) upstream of the transcription factor Scratch2 (Scrt2) locus with a predicted heterodimerization motif for ASCL1 and POU3F2. In this study, we investigated the role of ASCL1 and this enhancer in regulating the expression of the Scrt2 in the embryonic spinal cord. We confirmed that cE1 region interacted with the Scrt2 promoter. cE1 was sufficient to mediate ASCL1-driven expression in the neural tube through the heterodimerization sites. Moreover, Scrt2 expression was inhibited when we removed cE1 from the genome. These findings strongly indicate that ASCL1 regulates Scrt2 transcription in the neural tube through cE1.

3.
Nat Commun ; 15(1): 90, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167340

RESUMO

Embryonic cells exhibit diverse metabolic states. Recent studies have demonstrated that metabolic reprogramming drives changes in cell identity by affecting gene expression. However, the connection between cellular metabolism and gene expression remains poorly understood. Here we report that glycolysis-regulated histone lactylation couples the metabolic state of embryonic cells with chromatin organization and gene regulatory network (GRN) activation. We found that lactylation marks genomic regions of glycolytic embryonic tissues, like the neural crest (NC) and pre-somitic mesoderm. Histone lactylation occurs in the loci of NC genes as these cells upregulate glycolysis. This process promotes the accessibility of active enhancers and the deployment of the NC GRN. Reducing the deposition of the mark by targeting LDHA/B leads to the downregulation of NC genes and the impairment of cell migration. The deposition of lactyl-CoA on histones at NC enhancers is supported by a mechanism that involves transcription factors SOX9 and YAP/TEAD. These findings define an epigenetic mechanism that integrates cellular metabolism with the GRNs that orchestrate embryonic development.


Assuntos
Redes Reguladoras de Genes , Histonas , Histonas/genética , Histonas/metabolismo , Fatores de Transcrição/metabolismo , Desenvolvimento Embrionário/genética , Mesoderma/metabolismo
4.
Semin Cell Dev Biol ; 138: 28-35, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35787974

RESUMO

The neural crest is a vertebrate-specific embryonic stem cell population that gives rise to a vast array of cell types throughout the animal body plan. These cells are first born at the edges of the central nervous system, from which they migrate extensively and differentiate into multiple cellular derivatives. Given the unique set of structures these cells comprise, the origin of the neural crest is thought to have important implications for the evolution and diversification of the vertebrate clade. In jawed vertebrates, neural crest cells exist as distinct subpopulations along the anterior-posterior axis. These subpopulations differ in terms of their respective differentiation potential and cellular derivatives. Thus, the modern neural crest is characterized as multipotent, migratory, and regionally segregated throughout the embryo. Here, we retrace the evolutionary origins of the neural crest, from the appearance of conserved regulatory circuitry in basal chordates to the emergence of neural crest subpopulations in higher vertebrates. Finally, we discuss a stepwise trajectory by which these cells may have arisen and diversified throughout vertebrate evolution.


Assuntos
Evolução Biológica , Crista Neural , Animais , Vertebrados/genética , Diferenciação Celular/fisiologia
5.
Methods Mol Biol ; 2599: 49-58, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36427142

RESUMO

In vitro differentiation systems provide a flexible platform for understanding complex developmental processes. Here, we provide a comprehensive protocol for the preparation and analysis of ChIP-seq libraries for human-induced neural crest cells (hiNCCs) from human embryonic stem cells (hESCs). This workflow is aimed at identifying interactions between transcription factors and cis regulatory elements, which serve as useful assays in uncovering gene regulatory principles during development.


Assuntos
Sequenciamento de Cromatina por Imunoprecipitação , Células-Tronco Embrionárias Humanas , Humanos , Imunoprecipitação da Cromatina/métodos , Diferenciação Celular , Sequências Reguladoras de Ácido Nucleico
6.
Dev Cell ; 57(19): 2257-2272.e5, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36182685

RESUMO

Yamanaka factors are essential for establishing pluripotency in embryonic stem cells, but their function in multipotent stem cell populations is poorly understood. Here, we show that OCT4 and SOX2 cooperate with tissue-specific transcription factors to promote neural crest formation. By assessing avian and human neural crest cells at distinct developmental stages, we characterized the epigenomic changes that occur during their specification, migration, and early differentiation. This analysis determined that the OCT4-SOX2 dimer is required to establish a neural crest epigenomic signature that is lost upon cell fate commitment. The OCT4-SOX2 genomic targets in the neural crest differ from those of embryonic stem cells, indicating the dimer displays context-specific functions. Binding of OCT4-SOX2 to neural crest enhancers requires pioneer factor TFAP2A, which physically interacts with the dimer to modify its genomic targets. Our results demonstrate how Yamanaka factors are repurposed in multipotent cells to control chromatin organization and define their developmental potential.


Assuntos
Crista Neural , Fator 3 de Transcrição de Octâmero , Diferenciação Celular , Cromatina/metabolismo , Epigenômica , Humanos , Crista Neural/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/metabolismo
7.
Elife ; 112022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36111787

RESUMO

Neuronal identity dictates the position in an epithelium, and the ability to detect, process, and transmit specific signals to specified targets. Transcription factors (TFs) determine cellular identity via direct modulation of genetic transcription and recruiting chromatin modifiers. However, our understanding of the mechanisms that define neuronal identity and their magnitude remain a critical barrier to elucidate the etiology of congenital and neurodegenerative disorders. The rodent vomeronasal organ provides a unique system to examine in detail the molecular mechanisms underlying the differentiation and maturation of chemosensory neurons. Here, we demonstrated that the identity of postmitotic/maturing vomeronasal sensory neurons (VSNs), and vomeronasal-dependent behaviors can be reprogrammed through the rescue of Tfap2e/AP-2ε expression in the Tfap2eNull mice, and partially reprogrammed by inducing ectopic Tfap2e expression in mature apical VSNs. We suggest that the TF Tfap2e can reprogram VSNs bypassing cellular plasticity restrictions, and that it directly controls the expression of batteries of vomeronasal genes.


Assuntos
Órgão Vomeronasal , Animais , Cromatina/metabolismo , Camundongos , Camundongos Knockout , Células Receptoras Sensoriais/metabolismo , Fatores de Transcrição/metabolismo , Órgão Vomeronasal/metabolismo
8.
FEBS J ; 289(17): 5166-5175, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34310060

RESUMO

Early embryogenesis requires the establishment of fields of progenitor cells with distinct molecular signatures. A balance of intrinsic and extrinsic cues determines the boundaries of embryonic territories and pushes progenitor cells toward different fates. This process involves multiple layers of regulation, including signaling systems, transcriptional networks, and post-transcriptional control. In recent years, microRNAs (miRNAs) have emerged as undisputed regulators of developmental processes. Here, we discuss how miRNAs regulate pattern formation during vertebrate embryogenesis. We survey how miRNAs modulate the activity of signaling pathways to optimize transcriptional responses in embryonic cells. We also examine how localized RNA interference can generate spatial complexity during early development. Unraveling the complex crosstalk between miRNAs, signaling systems and cell fate decisions will be crucial for our understanding of developmental outcomes and disease.


Assuntos
MicroRNAs , Animais , Redes Reguladoras de Genes , MicroRNAs/genética , MicroRNAs/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Vertebrados/genética , Vertebrados/metabolismo
9.
Dev Cell ; 56(9): 1268-1282.e6, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33852891

RESUMO

Cell fate commitment is controlled by cis-regulatory elements often located in remote regions of the genome. To examine the role of long-range DNA interactions in early development, we generated a high-resolution contact map of active enhancers in avian neural crest cells. This analysis uncovered a diverse repertoire of enhancers that are part of the gene regulatory network underlying specification. We found that neural crest identity is largely regulated by cis-regulatory elements that propagate signaling inputs to network components. These genomic sensors display a combination of optimal and suboptimal TCF/LEF-binding sites, which allow cells to respond to Wnt signaling in a position-dependent manner. We propose that, rather than acting as upstream activators, signaling systems feed into regulatory circuits in a hub-and-spoke architecture. These results shed light on the tridimensional organization of the neural crest genome and define how signaling systems provide progenitors with spatial cues that transform their molecular identity.


Assuntos
Conectoma , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/metabolismo , Transdução de Sinais/genética , Animais , Sítios de Ligação , Núcleo Celular/metabolismo , Embrião de Galinha , Cromatina/metabolismo , Redes Reguladoras de Genes , Modelos Biológicos , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Via de Sinalização Wnt
10.
Dev Biol ; 475: 245-255, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33548210

RESUMO

The neural crest is a migratory stem cell population that contributes to various tissues and organs during vertebrate embryonic development. These cells possess remarkable developmental plasticity and give rise to many different cell types, including chondrocytes, osteocytes, peripheral neurons, glia, melanocytes, and smooth muscle cells. Although the genetic mechanisms underlying neural crest development have been extensively studied, many facets of this process remain unexplored. One key aspect of cellular physiology that has gained prominence in the context of embryonic development is metabolic regulation. Recent discoveries in neural crest biology suggest that metabolic regulation may play a central role in the formation, migration, and differentiation of these cells. This possibility is further supported by clinical studies that have demonstrated a high prevalence of neural crest anomalies in babies with congenital metabolic disorders. Here, we examine why neural crest development is prone to metabolic disruption and discuss how carbon metabolism regulates developmental processes like epithelial-to-mesenchymal transition (EMT) and cell migration. Finally, we explore how understanding neural crest metabolism may inform upon the etiology of several congenital birth defects.


Assuntos
Desenvolvimento Embrionário/fisiologia , Crista Neural/citologia , Crista Neural/embriologia , Animais , Carbono/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicólise/fisiologia , Humanos , Crista Neural/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Vertebrados/embriologia
11.
PLoS Genet ; 17(1): e1009296, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33465092

RESUMO

The process of cell fate commitment involves sequential changes in the gene expression profiles of embryonic progenitors. This is exemplified in the development of the neural crest, a migratory stem cell population derived from the ectoderm of vertebrate embryos. During neural crest formation, cells transition through distinct transcriptional states in a stepwise manner. The mechanisms underpinning these shifts in cell identity are still poorly understood. Here we employ enhancer analysis to identify a genetic sub-circuit that controls developmental transitions in the nascent neural crest. This sub-circuit links Wnt target genes in an incoherent feedforward loop that controls the sequential activation of genes in the neural crest lineage. By examining the cis-regulatory apparatus of Wnt effector gene AXUD1, we found that multipotency factor SP5 directly promotes neural plate border identity, while inhibiting premature expression of specification genes. Our results highlight the importance of repressive interactions in the neural crest gene regulatory network and illustrate how genes activated by the same upstream signal become temporally segregated during progressive fate restriction.


Assuntos
Elementos Facilitadores Genéticos/genética , Crista Neural/crescimento & desenvolvimento , Placa Neural/crescimento & desenvolvimento , Fatores de Transcrição/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Diferenciação Celular/genética , Movimento Celular/genética , Embrião de Galinha , Proteínas de Ligação a DNA/genética , Ectoderma/crescimento & desenvolvimento , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Humanos , Hibridização In Situ , Crista Neural/metabolismo , Placa Neural/metabolismo , Via de Sinalização Wnt/genética
12.
Proc Natl Acad Sci U S A ; 117(52): 33305-33316, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33376218

RESUMO

Ectodermal patterning is required for the establishment of multiple components of the vertebrate body plan. Previous studies have demonstrated that precise combinations of extracellular signals induce distinct ectodermal cell populations, such as the neural crest and the neural plate. Yet, we still lack understanding of how the response to inductive signals is modulated to generate the proper transcriptional output in target cells. Here we show that posttranscriptional attenuation of fibroblast growth factor (FGF) signaling is essential for the establishment of the neural crest territory. We found that neural crest progenitors display elevated expression of DICER, which promotes enhanced maturation of a set of cell-type-specific miRNAs. These miRNAs collectively target components of the FGF signaling pathway, a central player in the process of neural induction in amniotes. Inactivation of this posttranscriptional circuit results in a fate switch, in which neural crest cells are converted into progenitors of the central nervous system. Thus, the posttranscriptional attenuation of signaling systems is a prerequisite for proper segregation of ectodermal cell types. These findings demonstrate how posttranscriptional repression may alter the activity of signaling systems to generate distinct spatial domains of progenitor cells.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Crista Neural/metabolismo , Transdução de Sinais , Transcrição Gênica , Animais , Linhagem da Célula , Embrião de Galinha , Ectoderma/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Crista Neural/citologia , Ribonuclease III/metabolismo , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
Front Cell Dev Biol ; 8: 769, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32984310

RESUMO

Scratch2 is a transcription factor expressed in a very restricted population of vertebrate embryonic neural cell precursors involved in their survival, differentiation, and migration. The mechanisms that control its expression remain unknown and could contribute towards our understanding of gene regulation during neural differentiation and evolution. Here we investigate the role of microRNAs (miRNAs) in the Scrt2 post-transcriptional regulatory mechanism. We identified binding sites for miR-125b and -200b in the Scrt2 3'UTR in silico. We confirmed the repressive-mediated activity of the Scrt2 3'UTR through electroporation of luciferase constructs into chick embryos. Further, both CRISPR/Cas9-mediated deletion of miR-125b/-200b responsive elements from chicken Scrt2 3'UTR and expression of miRNAs sponges increased Scrt2 expression field, suggesting a role for these miRNAs as post-transcriptional regulators of Scrt2. The biological effect of miR-125b titration was much more pronounced than that of miR-200b. Therefore, we propose that, after transcription, miR-125b fine-tunes the Scrt2 expression domain.

14.
Dev Cell ; 53(2): 199-211.e6, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32243782

RESUMO

The Warburg effect is one of the metabolic hallmarks of cancer cells, characterized by enhanced glycolysis even under aerobic conditions. This physiological adaptation is associated with metastasis , but we still have a superficial understanding of how it affects cellular processes during embryonic development. Here we report that the neural crest, a migratory stem cell population in vertebrate embryos, undergoes an extensive metabolic remodeling to engage in aerobic glycolysis prior to delamination. This increase in glycolytic flux promotes Yap/Tead signaling, which activates the expression of a set of transcription factors to drive epithelial-to-mesenchymal transition. Our results demonstrate how shifts in carbon metabolism can trigger the gene regulatory circuits that control complex cell behaviors. These findings support the hypothesis that the Warburg effect is a precisely regulated developmental mechanism that is anomalously reactivated during tumorigenesis and metastasis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Glicólise , Crista Neural/fisiologia , Fatores de Transcrição/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Embrião de Galinha , Galinhas , Especificidade de Órgãos , Fatores de Transcrição/genética
15.
Genome Res ; 30(1): 35-48, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31848212

RESUMO

Cell fate commitment involves the progressive restriction of developmental potential. Recent studies have shown that this process requires not only shifts in gene expression but also an extensive remodeling of the epigenomic landscape. To examine how chromatin states are reorganized during cellular specification in an in vivo system, we examined the function of pioneer factor TFAP2A at discrete stages of neural crest development. Our results show that TFAP2A activates distinct sets of genomic regions during induction of the neural plate border and specification of neural crest cells. Genomic occupancy analysis revealed that the repertoire of TFAP2A targets depends upon its dimerization with paralogous proteins TFAP2C and TFAP2B. During gastrula stages, TFAP2A/C heterodimers activate components of the neural plate border induction program. As neurulation begins, TFAP2A trades partners, and TFAP2A/B heterodimers reorganize the epigenomic landscape of progenitor cells to promote neural crest specification. We propose that this molecular switch acts to drive progressive cell commitment, remodeling the epigenomic landscape to define the presumptive neural crest. Our findings show how pioneer factors regulate distinct genomic targets in a stage-specific manner and highlight how paralogy can serve as an evolutionary strategy to diversify the function of the regulators that control embryonic development.


Assuntos
Desenvolvimento Embrionário/genética , Epigênese Genética , Crista Neural/embriologia , Crista Neural/metabolismo , Multimerização Proteica , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Animais , Diferenciação Celular , Embrião de Galinha , Epigenômica , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Sequências Reguladoras de Ácido Nucleico , Fator de Transcrição AP-2/química
16.
Wiley Interdiscip Rev Syst Biol Med ; 12(2): e1468, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31702881

RESUMO

The neural crest is an ectodermal cell population that gives rise to over 30 cell types during vertebrate embryogenesis. These stem cells are formed at the border of the developing central nervous system and undergo extensive migration before differentiating into components of multiple tissues and organs. Neural crest formation and differentiation is a multistep process, as these cells transition through sequential regulatory states before adopting their adult phenotype. Such changes are governed by a complex gene regulatory network (GRN) that integrates environmental and cell-intrinsic inputs to regulate cell identity. Studies of neural crest cells in a variety of vertebrate models have elucidated the function and regulation of dozens of the molecular players that are part of this network. The neural crest GRN has served as a platform to explore the molecular control of multipotency, cell differentiation, and the evolution of vertebrates. In this review, we employ this genetic program as a stepping-stone to explore the architecture and the regulatory principles of developmental GRNs. We also discuss how modern genomic approaches can further expand our understanding of genetic networks in this system and others. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Biological Mechanisms > Cell Fates Developmental Biology > Lineages Models of Systems Properties and Processes > Cellular Models.


Assuntos
Redes Reguladoras de Genes , Crista Neural/metabolismo , Animais , Evolução Biológica , Diferenciação Celular , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Transdução de Sinais
17.
Nature ; 574(7780): 675-678, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31645763

RESUMO

The neural crest, an embryonic stem-cell population, is a vertebrate innovation that has been proposed to be a key component of the 'new head', which imbued vertebrates with predatory behaviour1,2. Here, to investigate how the evolution of neural crest cells affected the vertebrate body plan, we examined the molecular circuits that control neural crest development along the anteroposterior axis of a jawless vertebrate, the sea lamprey. Gene expression analysis showed that the cranial subpopulation of the neural crest of the lamprey lacks most components of a transcriptional circuit that is specific to the cranial neural crest in amniotes and confers the ability to form craniofacial cartilage onto non-cranial neural crest subpopulations3. Consistent with this, hierarchical clustering analysis revealed that the transcriptional profile of the lamprey cranial neural crest is more similar to the trunk neural crest of amniotes. Notably, analysis of the cranial neural crest in little skate and zebrafish embryos demonstrated that the transcriptional circuit that is specific to the cranial neural crest emerged via the gradual addition of network components to the neural crest of gnathostomes, which subsequently became restricted to the cephalic region. Our results indicate that the ancestral neural crest at the base of the vertebrate lineage possessed a trunk-like identity. We propose that the emergence of the cranial neural crest, by progressive assembly of an axial-specific regulatory circuit, allowed the elaboration of the new head during vertebrate evolution.


Assuntos
Evolução Biológica , Padronização Corporal , Cabeça , Crista Neural , Animais , Regulação da Expressão Gênica no Desenvolvimento , Cabeça/fisiologia , Lampreias/embriologia , Crista Neural/embriologia , Crista Neural/fisiologia , Crânio/embriologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética
18.
Methods Mol Biol ; 1920: 99-110, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30737688

RESUMO

The chick embryo is a powerful model for experimental embryology due to its accessibility, sturdiness, and ease of manipulation. Here we describe protocols for analysis of protein-DNA and protein-protein interactions in tissues and cells isolated from the developing chick. These assays are aimed at the identification of interactions between transcription factors and regulatory elements in the genome, and, in combination with functional assays, can be used for the delineation of gene regulatory circuits.


Assuntos
Aves , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Animais , Embrião de Galinha , Imunoprecipitação da Cromatina , Sequenciamento de Nucleotídeos em Larga Escala , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas
19.
Elife ; 72018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30520734

RESUMO

A crucial step in cell differentiation is the silencing of developmental programs underlying multipotency. While much is known about how lineage-specific genes are activated to generate distinct cell types, the mechanisms driving suppression of stemness are far less understood. To address this, we examined the regulation of the transcriptional network that maintains progenitor identity in avian neural crest cells. Our results show that a regulatory circuit formed by Wnt, Lin28a and let-7 miRNAs controls the deployment and the subsequent silencing of the multipotency program in a position-dependent manner. Transition from multipotency to differentiation is determined by the topological relationship between the migratory cells and the dorsal neural tube, which acts as a Wnt-producing stem cell niche. Our findings highlight a mechanism that rapidly silences complex regulatory programs, and elucidate how transcriptional networks respond to positional information during cell differentiation.


Assuntos
Proteínas Aviárias/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Crista Neural/metabolismo , Neurônios/metabolismo , Proteínas Wnt/genética , Dedos de Zinco/genética , Animais , Proteínas Aviárias/metabolismo , Diferenciação Celular , Movimento Celular , Embrião de Galinha , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , MicroRNAs/metabolismo , Morfolinos/genética , Morfolinos/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Neurônios/citologia , Neuropeptídeos/biossíntese , Neuropeptídeos/genética , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Transcrição Gênica , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
20.
Dev Biol ; 444 Suppl 1: S170-S180, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30071217

RESUMO

The neural crest is a migratory cell population that contributes to multiple tissues and organs during vertebrate embryonic development. It is remarkable in its ability to differentiate into an array of different cell types, including melanocytes, cartilage, bone, smooth muscle, and peripheral nerves. Although neural crest cells are formed along the entire anterior-posterior axis of the developing embryo, they can be divided into distinct subpopulations based on their axial level of origin. These groups of cells, which include the cranial, vagal, trunk, and sacral neural crest, display varied migratory patterns and contribute to multiple derivatives. While these subpopulations have been shown to be mostly plastic and to differentiate according to environmental cues, differences in their intrinsic potentials have also been identified. For instance, the cranial neural crest is unique in its ability to give rise to cartilage and bone. Here, we examine the molecular features that underlie such developmental restrictions and discuss the hypothesis that distinct gene regulatory networks operate in these subpopulations. We also consider how reconstructing the phylogeny of the trunk and cranial neural crest cells impacts our understanding of vertebrate evolution.


Assuntos
Crista Neural/embriologia , Crista Neural/metabolismo , Crista Neural/fisiologia , Animais , Evolução Biológica , Padronização Corporal/fisiologia , Cartilagem , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Desenvolvimento Embrionário , Redes Reguladoras de Genes , Humanos , Melanócitos , Tubo Neural , Neurogênese , Crânio , Vertebrados/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...